Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Phytother Res ; 38(5): 2496-2517, 2024 May.
Article in English | MEDLINE | ID: mdl-38447978

ABSTRACT

We investigated the mechanism by which quercetin preserves mitochondrial quality control (MQC) in cardiomyocytes subjected to ischemia-reperfusion stress. An enzyme-linked immunosorbent assay was employed in the in vivo experiments to assess myocardial injury markers, measure the transcript levels of SIRT5/DNAPK-cs/MLKL during various time intervals of ischemia-reperfusion, and observe structural changes in cardiomyocytes using transmission electron microscopy. In in vitro investigations, adenovirus transfection was employed to establish a gene-modified model of DNA-PKcs, and primary cardiomyocytes were obtained from a mouse model with modified SIRT5 gene. Reverse transcription polymerase chain reaction, laser confocal microscopy, immunofluorescence localization, JC-1 fluorescence assay, Seahorse energy analysis, and various other assays were applied to corroborate the regulatory influence of quercetin on the MQC network in cardiomyocytes after ischemia-reperfusion. In vitro experiments demonstrated that ischemia-reperfusion injury caused changes in the structure of the myocardium. It was seen that quercetin had a beneficial effect on the myocardial tissue, providing protection. As the ischemia-reperfusion process continued, the levels of DNA-PKcs/SIRT5/MLKL transcripts were also found to change. In vitro investigations revealed that quercetin mitigated cardiomyocyte injury caused by mitochondrial oxidative stress through DNA-PKcs, and regulated mitophagy and mitochondrial kinetics to sustain optimal mitochondrial energy metabolism levels. Quercetin, through SIRT5 desuccinylation, modulated the stability of DNA-PKcs, and together they regulated the "mitophagy-unfolded protein response." This preserved the integrity of mitochondrial membrane and genome, mitochondrial dynamics, and mitochondrial energy metabolism. Quercetin may operate synergistically to oversee the regulation of mitophagy and the unfolded protein response through DNA-PKcs-SIRT5 interaction.


Subject(s)
Myocytes, Cardiac , Quercetin , Sirtuins , Quercetin/pharmacology , Animals , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Mice , Sirtuins/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Oxidative Stress/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , DNA-Activated Protein Kinase/metabolism , Male , Mice, Inbred C57BL , Mitophagy/drug effects
2.
Nat Cancer ; 4(2): 181-202, 2023 02.
Article in English | MEDLINE | ID: mdl-36732634

ABSTRACT

Despite producing a panoply of potential cancer-specific targets, the proteogenomic characterization of human tumors has yet to demonstrate value for precision cancer medicine. Integrative multi-omics using a machine-learning network identified master kinases responsible for effecting phenotypic hallmarks of functional glioblastoma subtypes. In subtype-matched patient-derived models, we validated PKCδ and DNA-PK as master kinases of glycolytic/plurimetabolic and proliferative/progenitor subtypes, respectively, and qualified the kinases as potent and actionable glioblastoma subtype-specific therapeutic targets. Glioblastoma subtypes were associated with clinical and radiomics features, orthogonally validated by proteomics, phospho-proteomics, metabolomics, lipidomics and acetylomics analyses, and recapitulated in pediatric glioma, breast and lung squamous cell carcinoma, including subtype specificity of PKCδ and DNA-PK activity. We developed a probabilistic classification tool that performs optimally with RNA from frozen and paraffin-embedded tissues, which can be used to evaluate the association of therapeutic response with glioblastoma subtypes and to inform patient selection in prospective clinical trials.


Subject(s)
DNA-Activated Protein Kinase , Glioblastoma , Protein Kinase C-delta , Humans , DNA-Activated Protein Kinase/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , Multiomics , Protein Kinase C-delta/genetics , Proteomics
3.
Mol Cancer Ther ; 20(10): 1880-1892, 2021 10.
Article in English | MEDLINE | ID: mdl-34376581

ABSTRACT

As a conserved molecular chaperone, heat shock protein 90 (Hsp90) maintains the stability and homeostasis of oncoproteins and helps cancer cells survive. DNA-dependent protein kinase catalytic subunit (DNA-PKcs) plays a pivotal role in the non-homologous end joining pathway for DNA double-strand breaks (DSB) repair. Tumor cells contain higher levels of DNA-PKcs to survive by the hostile tumor microenvironment and various antitumor therapies. Here, we showed that increased levels of Hsp90α, Hsp90ß, and DNA-PKcs correlated with a poor overall survival in hepatocellular carcinoma (HCC). We revealed that Hsp90 N-terminal domain and C-terminal domain have different effects on DNA-PKcs protein and mRNA levels. The stability of DNA-PKcs depended on Hsp90α N-terminal nucleotide binding domain. Transcription factor SP1 regulates the transcription of PRKDC (gene name of DNA-PKcs) and is a client protein of Hsp90. Inhibition of Hsp90 N-terminal by STA9090 decreased the location of Hsp90α in nucleus, Hsp90α-SP1 interaction, SP1 level, and the binding of Hsp90α/SP1 at the proximal promoter region of PRKDC Because hyperthermia induces DSBs with increases level of DNA-PKcs, combined STA9090 treatment with hyperthermia effectively delayed the tumor growth and significantly decreased DNA-PKcs levels in xenografts model. Consistently, inhibition of Hsp90 increased the number of heat shock-induced γ-H2AX foci and delayed the repair of DSBs. Altogether, our results suggest that Hsp90 inhibitor STA9090 decreases DNA-PKcs protein stability and PRKDC mRNA level, which provide a theoretical basis for the promising combination therapy of hyperthermia and Hsp90 inhibitor in HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , DNA Damage , DNA-Activated Protein Kinase/chemistry , Gene Expression Regulation, Neoplastic/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Hyperthermia, Induced/adverse effects , RNA, Messenger/genetics , Animals , Apoptosis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , DNA Repair , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prognosis , Protein Stability , Survival Rate , Triazoles , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
J Med Chem ; 64(17): 12723-12737, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34428039

ABSTRACT

Eukaryotes have evolved two major pathways to repair potentially lethal DNA double-strand breaks. Homologous recombination represents a precise, DNA-template-based mechanism available during the S and G2 cell cycle phase, whereas non-homologous end joining, which requires DNA-dependent protein kinase (DNA-PK), allows for fast, cell cycle-independent but less accurate DNA repair. Here, we report the discovery of BAY-8400, a novel selective inhibitor of DNA-PK. Starting from a triazoloquinoxaline, which had been identified as a hit from a screen for ataxia telangiectasia and Rad3-related protein (ATR) inhibitors with inhibitory activity against ATR, ATM, and DNA-PK, lead optimization efforts focusing on potency and selectivity led to the discovery of BAY-8400. In in vitro studies, BAY-8400 showed synergistic activity of DNA-PK inhibition with DNA damage-inducing targeted alpha therapy. Combination of PSMA-targeted thorium-227 conjugate BAY 2315497 treatment of human prostate tumor-bearing mice with BAY-8400 oral treatment increased antitumor efficacy, as compared to PSMA-targeted thorium-227 conjugate monotherapy.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , DNA-Activated Protein Kinase/metabolism , Gene Expression Regulation/drug effects , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation , DNA-Activated Protein Kinase/genetics , Drug Synergism , Drug Therapy, Combination , Hepatocytes/drug effects , Humans , Mice , Molecular Structure , Phosphatidylinositol 3-Kinases/genetics , Rats , Structure-Activity Relationship , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
5.
Cell Death Dis ; 11(7): 602, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32732911

ABSTRACT

Glioblastoma multiforme (GBM) is the most malignant primary brain tumor and has the highest mortality rate among cancers and high resistance to radiation and cytotoxic chemotherapy. Although some targeted therapies can partially inhibit oncogenic mutation-driven proliferation of GBM cells, therapies harnessing synthetic lethality are 'coincidental' treatments with high effectiveness in cancers with gene mutations, such as GBM, which frequently exhibits DNA-PKcs mutation. By implementing a highly efficient high-throughput screening (HTS) platform using an in-house-constructed genome-wide human microRNA inhibitor library, we demonstrated that miR-1193 inhibition sensitized GBM tumor cells with DNA-PKcs deficiency. Furthermore, we found that miR-1193 directly targets YY1AP1, leading to subsequent inhibition of FEN1, an important factor in DNA damage repair. Inhibition of miR-1193 resulted in accumulation of DNA double-strand breaks and thus increased genomic instability. RPA-coated ssDNA structures enhanced ATR checkpoint kinase activity, subsequently activating the CHK1/p53/apoptosis axis. These data provide a preclinical theory for the application of miR-1193 inhibition as a potential synthetic lethal approach targeting GBM cancer cells with DNA-PKcs deficiency.


Subject(s)
Brain Neoplasms/enzymology , Brain Neoplasms/genetics , DNA-Activated Protein Kinase/deficiency , Glioblastoma/enzymology , Glioblastoma/genetics , MicroRNAs/metabolism , Synthetic Lethal Mutations/genetics , Apoptosis , Ataxia Telangiectasia Mutated Proteins/metabolism , Base Sequence , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Checkpoint Kinase 1/metabolism , DNA Breaks, Double-Stranded , DNA-Activated Protein Kinase/metabolism , Flap Endonucleases/metabolism , Genomic Instability , Humans , MicroRNAs/genetics , Models, Biological , Reproducibility of Results , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , YY1 Transcription Factor/metabolism
6.
Biomed Pharmacother ; 129: 110427, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32574974

ABSTRACT

Triptolide is a multi-functional natural small molecular compound extracted from a traditional Chinese medicinal herb. Triptolide and its derivatives exhibit cytotoxicity through inducing DNA damage, therefore increasing sensitivity to DNA-damage based chemotherapy or radiotherapy in different types of cells. However, the regulatory mechanism of genotoxicity by triptolide, and the loss of genome integrity induced by triptolide are not fully understood. Here, we measured the effects of triptolide on genome integrity in a human fibroblast line HCA2-hTERT using the neutral comet assay. We demonstrated that treating cells with triptolide induced genomic instability in HCA2-hTERT cells. Furthermore, we observed the accumulation of γH2AX foci in triptolide treated cells than control cells at 24 h post ionizing radiation. Further mechanistic studies indicated that triptolide inhibited the enzymatic activity of DNA-PKcs, the critical nonhomologous end joining factor. In vitro kinase activity assays showed that triptolide suppressed the kinase activity of DNA-PKcs and molecular docking also predicted a potential interaction between triptolide and DNA-PKcs. As a consequence, we found that triptolide treatment enhanced the interaction between DNA-PKcs and KU80 and hampered the following recruitment of 53BP1. Altogether, our finding provides a new perspective about the toxicity of triptolide in non-cancer cells and highlights the necessity of taking genome effects of triptolide and its derivatives into consideration in the future clinical and research applications.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair/drug effects , DNA-Activated Protein Kinase/antagonists & inhibitors , Diterpenes/toxicity , Fibroblasts/drug effects , Genomic Instability/drug effects , Phenanthrenes/toxicity , Protein Kinase Inhibitors/pharmacology , Cell Line , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Epoxy Compounds/toxicity , Fibroblasts/enzymology , Fibroblasts/pathology , Histones/metabolism , Humans , Ku Autoantigen/metabolism , Phosphorylation , Telomerase/genetics , Telomerase/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism
7.
Nat Commun ; 9(1): 4342, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30337542

ABSTRACT

Oncolytic virotherapy is a promising therapeutic strategy that uses replication-competent viruses to selectively destroy malignancies. However, the therapeutic effect of certain oncolytic viruses (OVs) varies among cancer patients. Thus, it is necessary to overcome resistance to OVs through rationally designed combination strategies. Here, through an anticancer drug screening, we show that DNA-dependent protein kinase (DNA-PK) inhibition sensitizes cancer cells to OV M1 and improves therapeutic effects in refractory cancer models in vivo and in patient tumour samples. Infection of M1 virus triggers the transcription of interferons (IFNs) and the activation of the antiviral response, which can be abolished by pretreatment of DNA-PK inhibitor (DNA-PKI), resulting in selectively enhanced replication of OV M1 within malignancies. Furthermore, DNA-PK inhibition promotes the DNA damage response induced by M1 virus, leading to increased tumour cell apoptosis. Together, our study identifies the combination of DNA-PKI and OV M1 as a potential treatment for cancers.


Subject(s)
Antiviral Agents/pharmacology , DNA Damage , DNA-Activated Protein Kinase/antagonists & inhibitors , Oncolytic Viruses/physiology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Combined Modality Therapy , DNA-Activated Protein Kinase/metabolism , Drug Evaluation, Preclinical , Endoplasmic Reticulum Stress/drug effects , Humans , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Oncolytic Virotherapy , Protein Kinase Inhibitors/pharmacology , Rats
8.
Cell Death Dis ; 8(11): e3167, 2017 11 09.
Article in English | MEDLINE | ID: mdl-29120412

ABSTRACT

MicroRNAs (miRNAs) have been suggested to repress transcription via binding the 3'-untranslated regions of mRNAs. However, the involvement and details of miRNA-mediated epigenetic regulation, particularly in targeting genomic DNA and mediating epigenetic regulation, remain largely uninvestigated. In the present study, transcription factor CCAAT/enhancer binding protein delta (CEBPD) was responsive to the anticancer drug bortezomib, a clinical and highly selective drug for leukemia treatment, and contributed to bortezomib-induced cell death. Interestingly, following the identification of CEBPD-induced miRNAs, we found that miR-744, miR-3154 and miR-3162 could target CpG islands in the 5'-flanking region of the CEBPD gene. We previously demonstrated that the Yin Yang 1 (YY1)/polycomb group (PcG) protein/DNA methyltransferase (DNMT) complex is important for CCAAT/enhancer binding protein delta (CEBPD) gene inactivation; we further found that Argonaute 2 (Ago2) interacts with YY1 and binds to the CEBPD promoter. The miRNA/Ago2/YY1/PcG group protein/DNMT complex linked the inactivation of CEBPD and genes adjacent to its 5'-flanking region, including protein kinase DNA-activated catalytic polypeptide (PRKDC), minichromosome maintenance-deficient 4 (MCM4) and ubiquitin-conjugating enzyme E2 variant 2 (UBE2V2), upon bortezomib treatment. Moreover, we revealed that miRNA binding is necessary for YY1/PcG group protein/DNMT complex-mediated epigenetic gene silencing and is associated with bortezomib-induced methylation on genomic DNA. The present study successfully characterized the interactions of the miRNA/Ago2/YY1/PcG group protein/DNMT complex and provided new insights for miRNA-mediated epigenetic regulation in bortezomib-induced leukemic cell arrest and cell death.


Subject(s)
Apoptosis/drug effects , Bortezomib/pharmacology , Leukemia/physiopathology , MicroRNAs/metabolism , 3' Untranslated Regions , Antineoplastic Agents/pharmacology , Argonaute Proteins/chemistry , Argonaute Proteins/metabolism , CCAAT-Enhancer-Binding Protein-delta/genetics , CCAAT-Enhancer-Binding Protein-delta/metabolism , Cell Line, Tumor , CpG Islands , DNA Methylation/drug effects , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Gene Silencing , Humans , Leukemia/metabolism , Ligases/genetics , Ligases/metabolism , MicroRNAs/genetics , Minichromosome Maintenance Complex Component 4/genetics , Minichromosome Maintenance Complex Component 4/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , Transcription, Genetic/drug effects , Ubiquitin-Conjugating Enzymes , YY1 Transcription Factor/chemistry , YY1 Transcription Factor/metabolism
9.
Oncotarget ; 7(40): 65504-65513, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27602767

ABSTRACT

Radiotherapy is based on the induction of lethal DNA damage, primarily DNA double-strand breaks (DSB). Efficient DSB repair via Non-Homologous End Joining or Homologous Recombination can therefore undermine the efficacy of radiotherapy. By suppressing DNA-DSB repair with hyperthermia (HT) and DNA-PKcs inhibitor NU7441 (DNA-PKcsi), we aim to enhance the effect of radiation.The sensitizing effect of HT for 1 hour at 42°C and DNA-PKcsi [1 µM] to radiation treatment was investigated in cervical and breast cancer cells, primary breast cancer sphere cells (BCSCs) enriched for cancer stem cells, and in an in vivo human tumor model. A significant radio-enhancement effect was observed for all cell types when DNA-PKcsi and HT were applied separately, and when both were combined, HT and DNA-PKcsi enhanced radio-sensitivity to an even greater extent. Strikingly, combined treatment resulted in significantly lower survival rates, 2 to 2.5 fold increase in apoptosis, more residual DNA-DSB 6 h post treatment and a G2-phase arrest. In addition, tumor growth analysis in vivo showed significant reduction in tumor growth and elevated caspase-3 activity when radiation was combined with HT and DNA-PKcsi compared to radiation alone. Importantly, no toxic side effects of HT or DNA-PKcsi were found.In conclusion, inhibiting DNA-DSB repair using HT and DNA-PKcsi before radiotherapy leads to enhanced cytotoxicity in cancer cells. This effect was even noticed in the more radio-resistant BCSCs, which are clearly sensitized by combined treatment. Therefore, the addition of HT and DNA-PKcsi to conventional radiotherapy is promising and might contribute to more efficient tumor control and patient outcome.


Subject(s)
Breast Neoplasms/therapy , Chromones/pharmacology , DNA-Activated Protein Kinase/antagonists & inhibitors , Hyperthermia, Induced , Morpholines/pharmacology , Neoplastic Stem Cells/radiation effects , Radiation-Sensitizing Agents/pharmacology , Uterine Cervical Neoplasms/therapy , Animals , Breast Neoplasms/pathology , DNA Breaks, Double-Stranded , DNA Damage , DNA End-Joining Repair , DNA Repair , Female , Homologous Recombination , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Radiation Tolerance , Radiotherapy , Tumor Cells, Cultured , Uterine Cervical Neoplasms/pathology
10.
Biomed Pharmacother ; 83: 693-703, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27470570

ABSTRACT

DNA-dependent protein kinase (DNA-PK) is a key enzyme in non-homologous DNA end joining (NHEJ) repair pathway. The targeted inhibition of such enzyme would furnish a valuable option for cancer treatment. In this study we report the development of validation of enzyme homology model, and the subsequent use of this model to perform docking-based virtual screening against a database of FDA-approved drugs. The nominated highest ranking hits (Praziquantel and Dutasteride) were subjected to biological investigation. Additionally, molecular dynamic study was carried-out for binding mode exploration. Results of the biological evaluation revealed that both compounds inhibit the DNA-PK enzymatic activity at relatively high concentration levels with an IC50 of 17.3µM for praziquantel and >20µM for dutasteride. Furthermore, both agents enhanced the anti-proliferative effects of doxorubicin and cisplatin on breast cancer (MCF7) and lung cancer (A549) cell lines. This result indicates that these two hits are good candidate as DNA-PK inhibitors and worth further structural modifications to enhance their enzyme inhibitory effects.


Subject(s)
Computer Simulation , DNA-Activated Protein Kinase/antagonists & inhibitors , Drug Evaluation, Preclinical , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Kinase Inhibitors/analysis , Protein Kinase Inhibitors/chemistry , Structural Homology, Protein , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Catalytic Domain , DNA-Activated Protein Kinase/chemistry , DNA-Activated Protein Kinase/metabolism , Dutasteride/chemistry , Dutasteride/pharmacology , Humans , Ligands , Praziquantel/chemistry , Praziquantel/pharmacology , Protein Kinase Inhibitors/pharmacology , ROC Curve
11.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(7): 940-4, 2016 Jul.
Article in Chinese | MEDLINE | ID: mdl-27363277

ABSTRACT

Objective To investigate the effect of evodiamine on the radiosensitivity of esophageal squamous cell cancer Eca-109 cells. Methods Eca-109 cells were treated with various concentrations of evodiamine [(10, 20, 40, 60, 80, 100, 120) µg/mL], and then cell proliferation was examined by MTT assay. After the optimal evodiamine concentration was determined, the cells were divided into radiation group (0, 2, 4, 6, 8 Gy X-ray radiation) and radiation combined with evodiamine group (80 µg/mL evodiamine and 0, 2, 4, 6, 8 Gy X-ray radiation) .The radiosensitivity of Eca-109 cells was detected using colony formation assay. Flow cytometry was used to determine cell cycle of Eca-109 cells. The protein expressions of Ku70, Ku80, DNA-PKcs and Rad51 were examined by Western blotting. Results MTT assay showed that evodiamine decreased the proliferation of Eca-109 cells in a concentration-dependent manner. The inhibition reached the maximal level at 80 µg/mL. Compared with radiotherapy alone, the combination of 80 µg/mL evodiamine and radiotherapy improved survival curve and decreased the values of D0 and Dq. Sensitizer enhancement ratio was 1.86±0.06. Furthermore, cell cycle analysis revealed that evodiamine suppressed radiotherapy-induced the G2/M arrest. Additionally, evodiamine treatment also significantly inhibited radiotherapy-induced increase in Ku70, Ku80, DNA-PKcs and Rad51 expressions. Conclusion Evodiamine enhances radiosensitivity of Eca-109 cells during radiotherapy. The effect may be associated with the inhibition of G2/M arrest and the attenuation of Ku70, Ku80, DNA-PKcs and Rad51 expressions.


Subject(s)
Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Quinazolines/pharmacology , Radiation Tolerance/drug effects , Blotting, Western , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , DNA-Activated Protein Kinase/metabolism , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Humans , Ku Autoantigen/metabolism , Nuclear Proteins/metabolism , Plant Extracts/pharmacology , Rad51 Recombinase/metabolism , X-Rays
12.
Chem Biol ; 22(7): 849-61, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26119999

ABSTRACT

Regulatory mechanisms underlying γH2AX induction and the associated cell fate decision during DNA damage response (DDR) remain obscure. Here, we discover a bromodomain (BRD)-like module in DNA-PKcs (DNA-PKcs-BRD) that specifically recognizes H2AX acetyl-lysine 5 (K5ac) for sequential induction of γH2AX and concurrent cell fate decision(s). First, top-down mass spectrometry of radiation-phenotypic, full-length H2AX revealed a radiation-inducible, K5ac-dependent induction of γH2AX. Combined approaches of sequence-structure modeling/docking, site-directed mutagenesis, and biochemical experiments illustrated that through docking on H2AX K5ac, this non-canonical BRD determines not only the H2AX-targeting activity of DNA-PKcs but also the over-activation of DNA-PKcs in radioresistant tumor cells, whereas a Kac antagonist, JQ1, was able to bind to DNA-PKcs-BRD, leading to re-sensitization of tumor cells to radiation. This study elucidates the mechanism underlying the H2AX-dependent regulation of DNA-PKcs in ionizing radiation-induced, differential DDR, and derives an unconventional, non-catalytic domain target in DNA-PKs for overcoming resistance during cancer radiotherapy.


Subject(s)
DNA Breaks, Double-Stranded , DNA-Activated Protein Kinase/chemistry , DNA-Activated Protein Kinase/metabolism , Histones/chemistry , Histones/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Amino Acid Sequence , Cell Line, Tumor , DNA Repair , DNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , K562 Cells , Lysine/metabolism , Molecular Sequence Data , Phosphorylation , Protein Structure, Tertiary , Radiation Tolerance
13.
Zhong Yao Cai ; 38(6): 1247-50, 2015 Jun.
Article in Chinese | MEDLINE | ID: mdl-26762068

ABSTRACT

OBJECTIVE: To study the inhibitory effect of Yiqi Chutan Recipe on the transplanted tumor through endoplasmic reticulum UPR-mediated approach. METHODS: 40 lung cancer A549 cells models transplanted in nude mice were established. On the 7th day of inoculation, mice were randomly divided into model group( saline group) , Cisplatin group (0.002 g/kg), Yiqi Chutan Recipe low dose group (3.0 g/kg), Yiqi Chutan Recipe high dose group(6. 0 g/kg)and Yiqi Chutan Recipe (3.0 g/kg)with Cisplatin group (0.002 g/kg). Each aforementioned group had eight mice. Mice were treated by Yiqi Chutan Recipe to gavage one time a day, for 21 days, and by Cisplatin Injection to intraperitoneal injection one time a day, for 7 days. On the 22th day, all mice were executed to death. Then each tumor's weight and volume were measured, and the expression of Caspase-4 and DNA-PK protein were detected through immunohistochemical method and Western blot method. RESULTS: Compared with model group, the tumors' volume and weight of Yiqi Chutan Recipe high dose group and Yiqi Chutan Recipe with Cisplatin group were decreased, but the expressions of Caspase-4 and DNA-PK protein in tumors were increased (P < 0.01). Yiqi Chutan Recipe with Cisplatin Group had the better effect (P < 0.05). CONCLUSION: Yiqi Chutan Recipe has a certain inhibitory effect on A549 lung cancer in mice and its possible mechanism is relevant to the increase of expression of Caspase-4 and DNA-PK protein.


Subject(s)
Apoptosis , Caspases, Initiator/metabolism , DNA-Activated Protein Kinase/metabolism , Drugs, Chinese Herbal/pharmacology , Lung Neoplasms/pathology , Nuclear Proteins/metabolism , Animals , Cell Line, Tumor , Humans , Lung Neoplasms/metabolism , Mice , Mice, Nude , Neoplasm Transplantation
14.
Mol Cell Biochem ; 399(1-2): 269-78, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25348361

ABSTRACT

The aim of the present study was to investigate the effects of DNA-PKcs deficiency on the chemosensitivity of human hepatoma HepG2 cells to cisplatin (CDDP) and 5-fluorouracil (5-Fu), and to explore the underlying molecular mechanism. After transfection with DNA-PKcs siRNA or control siRNA, HepG2 cells were exposed to combination treatment of CDDP and 5-Fu. The cell viability, DNA damage, cell apoptosis, intracellular reactive oxygen species and glutathione (GSH) level, expression of apoptosis related proteins, activity of phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway, and nuclear factor-κB (NF-κB) pathways were assessed. The combination of CDDP and 5-Fu had a synergistic cytotoxic effect in HepG2 cells in terms of the cell viability, DNA damage, apoptosis, and oxidative stress level. DNA-PKcs siRNA could sensitize the HepG2 cells to the combined treatment. DNA-PKcs suppression further reduced the Akt phosphorylation level and Bcl-2 expression in HepG2 cells exposed to CDDP and 5-Fu, but enhanced the expression of pro-apoptotic proteins p53 and caspase-3. Moreover, CDDP could inhibit the transcriptional activity of NF-κB through degradation of IkB-α, while 5-Fu alone seemed in some extent increases the NF-κB activity. The combined treatment with CDDP and 5-Fu resulted in significantly decrease of the transcriptional activity of NF-κB, which was further aggravated by DNA-PKcs siRNA treatment. In conclusion, DNA-PKcs suppression had complementary effects in combination with CDDP and 5-Fu treatment in HepG2 cells, which was associated with suppression of NF-κB signaling pathway cascade, activation of caspase-3 and p53, as well as down-regulation of Bcl-2 and GSH.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , DNA-Activated Protein Kinase/metabolism , Fluorouracil/pharmacology , Nuclear Proteins/metabolism , Apoptosis , Cell Survival/drug effects , DNA Damage , DNA-Activated Protein Kinase/genetics , Drug Resistance, Neoplasm , Gene Knockdown Techniques , Glutathione/metabolism , Hep G2 Cells , Humans , NF-kappa B/metabolism , Nuclear Proteins/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
15.
Salud colect ; 10(3): 407-415, sep.-dic. 2014.
Article in Spanish | LILACS | ID: lil-733299

ABSTRACT

La extensa obra de Javier Auyero sobre los sectores populares en América Latina inquieta por su complejidad sociológica y política. Alejada de los lugares comunes sobre cómo viven, sufren y se relacionan los habitantes de los márgenes de nuestras ciudades, su programa de veinte años de investigación aborda las consecuencias del neoliberalismo en la marginalidad urbana. Por la publicación de su último libro, Pacientes del Estado (2013), Salud Colectiva lo invita a reflexionar sobre las conexiones, no siempre observadas, entre la espera y la dominación política en oficinas estatales, escuelas y hospitales. Su estrategia etnográfica le permite ingresar sin prejuicios a un universo social atravesado por posicionamientos sociales polarizantes. En los encuentros cotidianos de los pobres con diversas formas de poder estatal, afirma, se reproducen prácticas -no todas ellas igualmente conscientes y planificadas- que imparten educación política y culminan convirtiendo a quienes deberían ser ciudadanos con derechos en pacientes del Estado.


The extensive work of Javier Auyero regarding the poor in Latin America is disturbing in its sociological and political complexity. Instead of falling into the commonplace explorations of how inhabitants at the margins of our cities live, suffer and relate, his twenty years of research have focused on the consequences of neoliberalism in urban marginality. In light of the publication of his last book Patients of the State (2013), Salud Colectiva invited Auyero to reflect on the connections, not always observed, between waiting and political domination in government offices, schools and hospitals. His ethnographic strategy allows him to enter without prejudices into a social universe marked by polarizing political positions. He affirms that in the everyday encounters of poor people with the diverse forms of state power, practices are reproduced - not all of which are equally conscious and planned - that impart a political education and end up turning those who should be citizens into patients of the State.


Subject(s)
Humans , Carcinoma, Hepatocellular , DNA-Binding Proteins , Hyperthermia, Induced , Liver Neoplasms , Protein Serine-Threonine Kinases/metabolism , Cell Survival/radiation effects , DNA Repair , DNA-Activated Protein Kinase , Nuclear Proteins , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Radiation Tolerance , Tumor Cells, Cultured
16.
Am J Chin Med ; 42(3): 729-42, 2014.
Article in English | MEDLINE | ID: mdl-24871662

ABSTRACT

Bufalin is a key component of a Chinese medicine (Chan Su) and has been proved effective in killing various cancer cells. Its role in inducing DNA damage and the inhibition of the DNA damage response (DDR) has been reported, but none have studied such action in lung cancer in detail. In this study, we demonstrated bufalin-induced DNA damage and condensation in NCI-H460 cells through a comet assay and DAPI staining, respectively. Western blotting indicated that bufalin suppressed the protein levels associated with DNA damage and repair, such as a DNA dependent serine/threonine protein kinase (DNA-PK), DNA repair proteins breast cancer 1, early onset (BRCA1), 14-3-3 σ (an important checkpoint keeper of DDR), mediator of DNA damage checkpoint 1 (MDC1), O6-methylguanine-DNA methyltransferase (MGMT) and p53 (tumor suppressor protein). Bufalin could activate phosphorylated p53 in NCI-H460 cells. DNA damage in NCI-H460 cells after treatment with bufalin up-regulated its ATM and ATR genes, which encode proteins functioning as sensors in DDR, and also up-regulated the gene expression (mRNA) of BRCA1 and DNA-PK. But bufalin suppressed the gene expression (mRNA) of p53 and 14-3-3 σ, however, bufalin did not significantly affect the mRNA of MGMT. In conclusion, bufalin induced DNA damage in NCI-H460 cells and also inhibited its DNA repair and checkpoint function.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Bufanolides/pharmacology , DNA Damage/drug effects , DNA Damage/genetics , DNA Repair/drug effects , Lung Neoplasms/genetics , Lung Neoplasms/pathology , 14-3-3 Proteins/metabolism , Adaptor Proteins, Signal Transducing , BRCA1 Protein/metabolism , Biomarkers, Tumor/metabolism , Cell Cycle Proteins , DNA Modification Methylases/metabolism , DNA Repair/genetics , DNA Repair Enzymes/metabolism , DNA-Activated Protein Kinase/metabolism , Exoribonucleases/metabolism , Genes, cdc/drug effects , Genes, cdc/genetics , Humans , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
17.
Environ Mol Mutagen ; 55(5): 436-48, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24500925

ABSTRACT

Radiation exposure is a serious threat to biomolecules, particularly DNA, proteins and lipids. Various exogenous substances have been reported to protect these biomolecules. In this study we explored the effect of pre-treatment with G-002M, a mixture of three active derivatives isolated from the rhizomes of Podophyllum hexandrum, on DNA damage response in irradiated human blood leukocytes. Blood was collected from healthy male volunteers, preincubated with G-002M and then irradiated with various doses of radiation. Samples were analyzed using flow cytometry to quantify DNA double strand break (DSB) biomarkers including γ-H2AX, P53BP1 and levels of ligase IV. Blood samples were irradiated in vitro and processed to determine time and dose-dependent kinetics. Semiquantitative RT-PCR was performed at various time points to measure gene expression of DNA-PKcs, Ku80, ATM, and 53BP1; each of these genes is involved in DNA repair signaling. Pre-treatment of blood with G-002M resulted in reduction of γ-H2AX and P53BP1 biomarkers levels and elevated ligase IV levels relative to non-G-002M-treated irradiated cells. These results confirm suppression in radiation-induced DNA DSBs. Samples pre-treated with G-002M and then irradiated also showed significant up-regulation of DNA-PKcs and Ku80 and downregulation of ATM and 53BP1 gene expressions, suggesting that G-002M plays a protective role against DNA damage. The protective effect of G-002M may be due to its ability to scavange radiation-induced free radicals or assist in DNA repair. Further studies are needed to decipher the role of G-002M on signaling molecules involved in radiation-induced DNA damage repair pathways.


Subject(s)
DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , Drugs, Chinese Herbal/pharmacology , Flavonoids/pharmacology , Gamma Rays/adverse effects , Leukocytes/drug effects , Podophyllum/chemistry , Radiation-Protective Agents/pharmacology , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Berberidaceae , Cells, Cultured , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Flavonoids/chemistry , Histones/genetics , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Ku Autoantigen , Leukocytes/metabolism , Leukocytes/radiation effects , Lymphocytes/drug effects , Lymphocytes/metabolism , Lymphocytes/radiation effects , Male , Monocytes/drug effects , Monocytes/metabolism , Monocytes/radiation effects , Phosphorylation/drug effects , Phosphorylation/radiation effects , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor p53-Binding Protein 1
18.
Cancer Immunol Immunother ; 62(9): 1485-97, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23817722

ABSTRACT

INTRODUCTION: Lung cancer is the most common cancer worldwide. Every year, as many people die of lung cancer as of breast, colon and rectum cancers combined. Because most patients are being diagnosed in advanced, not resectable stages and therefore have a poor prognosis, there is an urgent need for alternative therapies. Since it has been demonstrated that a high number of tumor- and stromal-infiltrating cytotoxic T cells (CTLs) is associated with an increased disease-specific survival in lung cancer patients, it can be assumed that immunotherapy, e.g. peptide vaccines that are able to induce a CTL response against the tumor, might be a promising approach. METHODS: We analyzed surgically resected lung cancer tissues with respect to HLA class I- and II-presented peptides and gene expression profiles, aiming at the identification of (novel) tumor antigens. In addition, we tested the ability of HLA ligands derived from such antigens to generate a CTL response in healthy donors. RESULTS: Among 170 HLA ligands characterized, we were able to identify several potential targets for specific CTL recognition and to generate CD8+ T cells which were specific for peptides derived from cyclin D1 or protein-kinase, DNA-activated, catalytic polypeptide and lysed tumor cells loaded with peptide. CONCLUSIONS: This is the first molecular analysis of HLA class I and II ligands ex vivo from human lung cancer tissues which reveals known and novel tumor antigens able to elicit a CTL response.


Subject(s)
Epitopes, T-Lymphocyte/immunology , HLA Antigens/immunology , Lung Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Cyclin D1/immunology , DNA-Activated Protein Kinase/immunology , Dendritic Cells/immunology , Gene Expression , Humans , Immunohistochemistry , Immunotherapy , Ligands , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Molecular Sequence Data , Nuclear Proteins/immunology , Peptides/immunology
19.
J Nutr Biochem ; 24(5): 781-7, 2013 May.
Article in English | MEDLINE | ID: mdl-22841545

ABSTRACT

Selenium induces a senescence response in cells through induction of ataxia-telangiectasia mutated (ATM) and reactive oxygen species (ROS). Although a role of the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) in DNA double-strand break repair is established, it is unclear how these proteins function in response to selenium-induced oxidative stress and senescence induction. In this study, we demonstrated that pretreating normal human diploid fibroblasts with DNA-PK kinase inhibitor NU 7026 suppressed selenium-induced senescence response. Selenium treatment induced phosphorylation of DNA-PKcs on Thr-2647 and Ser-2056, the extent of which was decreased in the presence of ATM kinase inhibitor KU 55933 or the antioxidants N-acetylcysteine or 2,2,6,6-tetramethylpiperidine-1-oxyl. In contrast, the selenium-induced phosphorylation of ATM on Ser-1981 was not affected by NU 7026. Cells deficient in DNA-PKcs or pretreated with NU 7026 or N-acetylcysteine were defective in selenite-induced ROS formation. Taken together, these results indicate a distinct role of DNA-PKcs, in which this kinase can respond to and feed forward selenium-induced ROS formation and is placed downstream of ATM in the resultant senescence response.


Subject(s)
Catalytic Domain , Cellular Senescence/drug effects , DNA-Activated Protein Kinase/metabolism , Oxidative Stress/drug effects , Selenium/pharmacology , Acetylcysteine/pharmacology , Antioxidants/pharmacology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cells, Cultured , Chromones/pharmacology , DNA Repair/drug effects , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/genetics , Dextrans/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Morpholines/pharmacology , Mutation , Phosphorylation , Pyrones/pharmacology , Reactive Oxygen Species/metabolism , Selenious Acid/pharmacology
20.
Biochim Biophys Acta ; 1833(1): 90-100, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23098854

ABSTRACT

Non-homologous end joining (NHEJ) is one of the major pathways that repairs double-stranded DNA breaks (DSBs). Activation of DNA-PK is required for NHEJ. However, the mechanism leading to DNA-PKcs activation remains incompletely understood. We provide evidence here that the MEK-ERK pathway plays a role in DNA-PKcs-mediated NHEJ. In comparison to the vehicle control (DMSO), etoposide (ETOP)-induced DSBs in MCF7 cells were more rapidly repaired in the presence of U0126, a specific MEK inhibitor, based on the reduction of γH2AX and tail moments. Additionally, U0126 increased reactivation of luciferase activity, which resulted from the repair of restriction enzyme-cleaved DSBs. Furthermore, while inhibition of ERK activation using the dominant-negative MEK1K97M accelerated the repair of DSBs, enforcing ERK activation with the constitutively active MEK1Q56P reduced DSB repair. In line with MEK activating ERK1 and ERK2 kinases, knockdown of either ERK1 or ERK2 increased DSB repair. Consistent with the activation of DNA-PKcs being required for NHEJ, we demonstrated that inhibition of ERK activation using U0126, MEK1K97M, and knockdown of ERK1 or ERK2 enhanced ETOP-induced activation of DNA-PKcs. Conversely, enforcing ERK activation by MEK1Q56P reduced ETOP-initiated DNA-PKcs activation. Taken together, we demonstrate that ERK reduces NHEJ-mediated repair of DSBs via attenuation of DNA-PKcs activation.


Subject(s)
DNA Breaks, Double-Stranded/drug effects , DNA End-Joining Repair/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Nuclear Proteins/agonists , RNA, Small Interfering/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , DNA-Activated Protein Kinase/metabolism , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Etoposide/pharmacology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Humans , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Nuclear Proteins/metabolism , Tumor Cells, Cultured , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL